Skip to main content
Top
Published in: European Journal of Medical Research 1/2023

Open Access 01-12-2023 | Crohn's Disease | Research

Predicting diagnostic biomarkers associated with immune infiltration in Crohn's disease based on machine learning and bioinformatics

Authors: Wenhui Bao, Lin Wang, Xiaoxiao Liu, Ming Li

Published in: European Journal of Medical Research | Issue 1/2023

Login to get access

Abstract

Objective

The objective of this study is to investigate potential biomarkers of Crohn's disease (CD) and the pathological importance of infiltration of associated immune cells in disease development using machine learning.

Methods

Three publicly accessible CD gene expression profiles were obtained from the GEO database. Inflammatory tissue samples were selected and differentiated between colonic and ileal tissues. To determine the differentially expressed genes (DEGs) between CD and healthy controls, the larger sample size was merged as a training unit. The function of DEGs was comprehended through disease enrichment (DO) and gene set enrichment analysis (GSEA) on DEGs. Promising biomarkers were identified using the support vector machine-recursive feature elimination and lasso regression models. To further clarify the efficacy of potential biomarkers as diagnostic genes, the area under the ROC curve was observed in the validation group. Additionally, using the CIBERSORT approach, immune cell fractions from CD patients were examined and linked with potential biomarkers.

Results

Thirty-four DEGs were identified in colon tissue, of which 26 were up-regulated and 8 were down-regulated. In ileal tissues, 50 up-regulated and 50 down-regulated DEGs were observed. Disease enrichment of colon and ileal DEGs primarily focused on immunity, inflammatory bowel disease, and related pathways. CXCL1, S100A8, REG3A, and DEFA6 in colon tissue and LCN2 and NAT8 in ileum tissue demonstrated excellent diagnostic value and could be employed as CD gene biomarkers using machine learning methods in conjunction with external dataset validation. In comparison to controls, antigen processing and presentation, chemokine signaling pathway, cytokine–cytokine receptor interactions, and natural killer cell-mediated cytotoxicity were activated in colonic tissues. Cytokine–cytokine receptor interactions, NOD-like receptor signaling pathways, and toll-like receptor signaling pathways were activated in ileal tissues. NAT8 was found to be associated with CD8 T cells, while CXCL1, S100A8, REG3A, LCN2, and DEFA6 were associated with neutrophils, indicating that immune cell infiltration in CD is closely connected.

Conclusion

CXCL1, S100A8, REG3A, and DEFA6 in colonic tissue and LCN2 and NAT8 in ileal tissue can be employed as CD biomarkers. Additionally, immune cell infiltration is crucial for CD development.
Literature
1.
go back to reference Dulai PS, Singh S, Vande Casteele N, Boland BS, Rivera-Nieves J, Ernst PB, et al. Should we divide Crohn’s disease into ileum-dominant and isolated colonic diseases? Clin Gastroenterol Hepatol. 2019;17(13):2634–43.PubMedPubMedCentralCrossRef Dulai PS, Singh S, Vande Casteele N, Boland BS, Rivera-Nieves J, Ernst PB, et al. Should we divide Crohn’s disease into ileum-dominant and isolated colonic diseases? Clin Gastroenterol Hepatol. 2019;17(13):2634–43.PubMedPubMedCentralCrossRef
2.
go back to reference Roda G, Chien Ng S, Kotze PG, Argollo M, Panaccione R, Spinelli A, et al. Crohn’s disease. Nat Rev Dis Primers. 2020;6(1):22.PubMedCrossRef Roda G, Chien Ng S, Kotze PG, Argollo M, Panaccione R, Spinelli A, et al. Crohn’s disease. Nat Rev Dis Primers. 2020;6(1):22.PubMedCrossRef
3.
go back to reference Garber A, Regueiro M. Extraintestinal manifestations of inflammatory bowel disease: epidemiology, etiopathogenesis, and management. Curr Gastroenterol Rep. 2019;21(7):31.PubMedCrossRef Garber A, Regueiro M. Extraintestinal manifestations of inflammatory bowel disease: epidemiology, etiopathogenesis, and management. Curr Gastroenterol Rep. 2019;21(7):31.PubMedCrossRef
4.
go back to reference Ng SC, Bernstein CN, Vatn MH, Lakatos PL, Loftus EV Jr, Tysk C, et al. Geographical variability and environmental risk factors in inflammatory bowel disease. Gut. 2013;62(4):630–49.PubMedCrossRef Ng SC, Bernstein CN, Vatn MH, Lakatos PL, Loftus EV Jr, Tysk C, et al. Geographical variability and environmental risk factors in inflammatory bowel disease. Gut. 2013;62(4):630–49.PubMedCrossRef
5.
go back to reference Molodecky NA, Soon IS, Rabi DM, Ghali WA, Ferris M, Chernoff G, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142(1):46–54.e42; quiz e30. Molodecky NA, Soon IS, Rabi DM, Ghali WA, Ferris M, Chernoff G, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142(1):46–54.e42; quiz e30.
6.
go back to reference Feuerstein JD, Cheifetz AS. Crohn disease: epidemiology, diagnosis, and management. Mayo Clin Proc. 2017;92(7):1088–103.PubMedCrossRef Feuerstein JD, Cheifetz AS. Crohn disease: epidemiology, diagnosis, and management. Mayo Clin Proc. 2017;92(7):1088–103.PubMedCrossRef
7.
go back to reference Ananthakrishnan AN, Bernstein CN, Iliopoulos D, Macpherson A, Neurath MF, Ali RAR, et al. Environmental triggers in IBD: a review of progress and evidence. Nat Rev Gastroenterol Hepatol. 2018;15(1):39–49.PubMedCrossRef Ananthakrishnan AN, Bernstein CN, Iliopoulos D, Macpherson A, Neurath MF, Ali RAR, et al. Environmental triggers in IBD: a review of progress and evidence. Nat Rev Gastroenterol Hepatol. 2018;15(1):39–49.PubMedCrossRef
8.
go back to reference Tsianos EV, Katsanos KH, Tsianos VE. Role of genetics in the diagnosis and prognosis of Crohn’s disease. World J Gastroenterol. 2011;17(48):5246–59.PubMedPubMedCentralCrossRef Tsianos EV, Katsanos KH, Tsianos VE. Role of genetics in the diagnosis and prognosis of Crohn’s disease. World J Gastroenterol. 2011;17(48):5246–59.PubMedPubMedCentralCrossRef
9.
go back to reference Danese S, Fiorino G, Fernandes C, Peyrin-Biroulet L. Catching the therapeutic window of opportunity in early Crohn’s disease. Curr Drug Targets. 2014;15(11):1056–63.PubMedCrossRef Danese S, Fiorino G, Fernandes C, Peyrin-Biroulet L. Catching the therapeutic window of opportunity in early Crohn’s disease. Curr Drug Targets. 2014;15(11):1056–63.PubMedCrossRef
10.
go back to reference Gajendran M, Loganathan P, Catinella AP, Hashash JG. A comprehensive review and update on Crohn’s disease. Disease-a-month : DM. 2018;64(2):20–57.CrossRef Gajendran M, Loganathan P, Catinella AP, Hashash JG. A comprehensive review and update on Crohn’s disease. Disease-a-month : DM. 2018;64(2):20–57.CrossRef
11.
go back to reference Torres J, Mehandru S, Colombel JF, Peyrin-Biroulet L. Crohn’s disease. Lancet (London, England). 2017;389(10080):1741–55.PubMedCrossRef Torres J, Mehandru S, Colombel JF, Peyrin-Biroulet L. Crohn’s disease. Lancet (London, England). 2017;389(10080):1741–55.PubMedCrossRef
12.
go back to reference Brazil JC, Louis NA, Parkos CA. The role of polymorphonuclear leukocyte trafficking in the perpetuation of inflammation during inflammatory bowel disease. Inflamm Bowel Dis. 2013;19(7):1556–65.PubMedCrossRef Brazil JC, Louis NA, Parkos CA. The role of polymorphonuclear leukocyte trafficking in the perpetuation of inflammation during inflammatory bowel disease. Inflamm Bowel Dis. 2013;19(7):1556–65.PubMedCrossRef
13.
go back to reference Marks DJ, Harbord MW, MacAllister R, Rahman FZ, Young J, Al-Lazikani B, et al. Defective acute inflammation in Crohn’s disease: a clinical investigation. Lancet (London, England). 2006;367(9511):668–78.PubMedCrossRef Marks DJ, Harbord MW, MacAllister R, Rahman FZ, Young J, Al-Lazikani B, et al. Defective acute inflammation in Crohn’s disease: a clinical investigation. Lancet (London, England). 2006;367(9511):668–78.PubMedCrossRef
14.
go back to reference Espaillat MP, Kew RR, Obeid LM. Sphingolipids in neutrophil function and inflammatory responses: mechanisms and implications for intestinal immunity and inflammation in ulcerative colitis. Adv Biol Regul. 2017;63:140–55.PubMedCrossRef Espaillat MP, Kew RR, Obeid LM. Sphingolipids in neutrophil function and inflammatory responses: mechanisms and implications for intestinal immunity and inflammation in ulcerative colitis. Adv Biol Regul. 2017;63:140–55.PubMedCrossRef
15.
go back to reference Ramos GP, Papadakis KA. Mechanisms of disease: inflammatory bowel diseases. Mayo Clin Proc. 2019;94(1):155–65.PubMedCrossRef Ramos GP, Papadakis KA. Mechanisms of disease: inflammatory bowel diseases. Mayo Clin Proc. 2019;94(1):155–65.PubMedCrossRef
16.
go back to reference Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy KM. Th17: an effector CD4 T cell lineage with regulatory T cell ties. Immunity. 2006;24(6):677–88.PubMedCrossRef Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy KM. Th17: an effector CD4 T cell lineage with regulatory T cell ties. Immunity. 2006;24(6):677–88.PubMedCrossRef
17.
go back to reference O’Connor W Jr, Zenewicz LA, Flavell RA. The dual nature of T(H)17 cells: shifting the focus to function. Nat Immunol. 2010;11(6):471–6.PubMedCrossRef O’Connor W Jr, Zenewicz LA, Flavell RA. The dual nature of T(H)17 cells: shifting the focus to function. Nat Immunol. 2010;11(6):471–6.PubMedCrossRef
18.
go back to reference Mayne CG, Williams CB. Induced and natural regulatory T cells in the development of inflammatory bowel disease. Inflamm Bowel Dis. 2013;19(8):1772–88.PubMedCrossRef Mayne CG, Williams CB. Induced and natural regulatory T cells in the development of inflammatory bowel disease. Inflamm Bowel Dis. 2013;19(8):1772–88.PubMedCrossRef
19.
go back to reference Fuss IJ, Neurath M, Boirivant M, Klein JS, de la Motte C, Strong SA, et al. Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn’s disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J Immunol. 1996;157(3):1261–70.PubMedCrossRef Fuss IJ, Neurath M, Boirivant M, Klein JS, de la Motte C, Strong SA, et al. Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn’s disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J Immunol. 1996;157(3):1261–70.PubMedCrossRef
20.
go back to reference Takayama T, Kamada N, Chinen H, Okamoto S, Kitazume MT, Chang J, et al. Imbalance of NKp44(+)NKp46(-) and NKp44(-)NKp46(+) natural killer cells in the intestinal mucosa of patients with Crohn's disease. Gastroenterology. 2010;139(3):882–92.e1–3. Takayama T, Kamada N, Chinen H, Okamoto S, Kitazume MT, Chang J, et al. Imbalance of NKp44(+)NKp46(-) and NKp44(-)NKp46(+) natural killer cells in the intestinal mucosa of patients with Crohn's disease. Gastroenterology. 2010;139(3):882–92.e1–3.
21.
go back to reference Torres J, Burisch J, Riddle M, Dubinsky M, Colombel JF. Preclinical disease and preventive strategies in IBD: perspectives, challenges and opportunities. Gut. 2016;65(7):1061–9.PubMedCrossRef Torres J, Burisch J, Riddle M, Dubinsky M, Colombel JF. Preclinical disease and preventive strategies in IBD: perspectives, challenges and opportunities. Gut. 2016;65(7):1061–9.PubMedCrossRef
22.
go back to reference Høivik ML, Moum B, Solberg IC, Henriksen M, Cvancarova M, Bernklev T. Work disability in inflammatory bowel disease patients 10 years after disease onset: results from the IBSEN Study. Gut. 2013;62(3):368–75.PubMedCrossRef Høivik ML, Moum B, Solberg IC, Henriksen M, Cvancarova M, Bernklev T. Work disability in inflammatory bowel disease patients 10 years after disease onset: results from the IBSEN Study. Gut. 2013;62(3):368–75.PubMedCrossRef
23.
go back to reference Frøslie KF, Jahnsen J, Moum BA, Vatn MH. Mucosal healing in inflammatory bowel disease: results from a Norwegian population-based cohort. Gastroenterology. 2007;133(2):412–22.PubMedCrossRef Frøslie KF, Jahnsen J, Moum BA, Vatn MH. Mucosal healing in inflammatory bowel disease: results from a Norwegian population-based cohort. Gastroenterology. 2007;133(2):412–22.PubMedCrossRef
24.
go back to reference Peyrin-Biroulet L, Harmsen WS, Tremaine WJ, Zinsmeister AR, Sandborn WJ, Loftus EV Jr. Surgery in a population-based cohort of Crohn’s disease from Olmsted County, Minnesota (1970–2004). Am J Gastroenterol. 2012;107(11):1693–701.PubMedPubMedCentralCrossRef Peyrin-Biroulet L, Harmsen WS, Tremaine WJ, Zinsmeister AR, Sandborn WJ, Loftus EV Jr. Surgery in a population-based cohort of Crohn’s disease from Olmsted County, Minnesota (1970–2004). Am J Gastroenterol. 2012;107(11):1693–701.PubMedPubMedCentralCrossRef
25.
go back to reference Louis E, Collard A, Oger AF, Degroote E, Aboul Nasr El Yafi FA, Belaiche J. Behaviour of Crohn's disease according to the Vienna classification: changing pattern over the course of the disease. Gut. 2001;49(6):777–82. Louis E, Collard A, Oger AF, Degroote E, Aboul Nasr El Yafi FA, Belaiche J. Behaviour of Crohn's disease according to the Vienna classification: changing pattern over the course of the disease. Gut. 2001;49(6):777–82.
26.
go back to reference Pierre N, Salée C, Vieujean S, Bequet E, Merli AM, Siegmund B, et al. Review article: distinctions between ileal and colonic Crohn’s disease: from physiology to pathology. Aliment Pharmacol Ther. 2021;54(6):779–91.PubMedCrossRef Pierre N, Salée C, Vieujean S, Bequet E, Merli AM, Siegmund B, et al. Review article: distinctions between ileal and colonic Crohn’s disease: from physiology to pathology. Aliment Pharmacol Ther. 2021;54(6):779–91.PubMedCrossRef
28.
go back to reference Odes S, Vardi H, Friger M, Wolters F, Hoie O, Moum B, et al. Effect of phenotype on health care costs in Crohn’s disease: a European study using the Montreal classification. J Crohns Colitis. 2007;1(2):87–96.PubMedCrossRef Odes S, Vardi H, Friger M, Wolters F, Hoie O, Moum B, et al. Effect of phenotype on health care costs in Crohn’s disease: a European study using the Montreal classification. J Crohns Colitis. 2007;1(2):87–96.PubMedCrossRef
29.
go back to reference Veauthier B, Hornecker JR. Crohn’s disease: diagnosis and management. Am Fam Physician. 2018;98(11):661–9.PubMed Veauthier B, Hornecker JR. Crohn’s disease: diagnosis and management. Am Fam Physician. 2018;98(11):661–9.PubMed
30.
go back to reference Peyrin-Biroulet L, Sandborn W, Sands BE, Reinisch W, Bemelman W, Bryant RV, et al. Selecting therapeutic targets in inflammatory bowel disease (STRIDE): determining therapeutic goals for treat-to-target. Am J Gastroenterol. 2015;110(9):1324–38.PubMedCrossRef Peyrin-Biroulet L, Sandborn W, Sands BE, Reinisch W, Bemelman W, Bryant RV, et al. Selecting therapeutic targets in inflammatory bowel disease (STRIDE): determining therapeutic goals for treat-to-target. Am J Gastroenterol. 2015;110(9):1324–38.PubMedCrossRef
31.
go back to reference Billiet T, Ferrante M, Van Assche G. The use of prognostic factors in inflammatory bowel diseases. Curr Gastroenterol Rep. 2014;16(11):416.PubMedCrossRef Billiet T, Ferrante M, Van Assche G. The use of prognostic factors in inflammatory bowel diseases. Curr Gastroenterol Rep. 2014;16(11):416.PubMedCrossRef
32.
go back to reference Allen PB, Gower-Rousseau C, Danese S, Peyrin-Biroulet L. Preventing disability in inflammatory bowel disease. Ther Adv Gastroenterol. 2017;10(11):865–76.CrossRef Allen PB, Gower-Rousseau C, Danese S, Peyrin-Biroulet L. Preventing disability in inflammatory bowel disease. Ther Adv Gastroenterol. 2017;10(11):865–76.CrossRef
35.
go back to reference Segal AW. The role of neutrophils in the pathogenesis of Crohn’s disease. Eur J Clin Invest. 2018;48(Suppl 2): e12983.PubMedCrossRef Segal AW. The role of neutrophils in the pathogenesis of Crohn’s disease. Eur J Clin Invest. 2018;48(Suppl 2): e12983.PubMedCrossRef
36.
go back to reference Smith AM, Rahman FZ, Hayee B, Graham SJ, Marks DJ, Sewell GW, et al. Disordered macrophage cytokine secretion underlies impaired acute inflammation and bacterial clearance in Crohn’s disease. J Exp Med. 2009;206(9):1883–97.PubMedPubMedCentralCrossRef Smith AM, Rahman FZ, Hayee B, Graham SJ, Marks DJ, Sewell GW, et al. Disordered macrophage cytokine secretion underlies impaired acute inflammation and bacterial clearance in Crohn’s disease. J Exp Med. 2009;206(9):1883–97.PubMedPubMedCentralCrossRef
37.
go back to reference Lipinski S, Till A, Sina C, Arlt A, Grasberger H, Schreiber S, et al. DUOX2-derived reactive oxygen species are effectors of NOD2-mediated antibacterial responses. J Cell Sci. 2009;122(Pt 19):3522–30.PubMedCrossRef Lipinski S, Till A, Sina C, Arlt A, Grasberger H, Schreiber S, et al. DUOX2-derived reactive oxygen species are effectors of NOD2-mediated antibacterial responses. J Cell Sci. 2009;122(Pt 19):3522–30.PubMedCrossRef
38.
go back to reference Schwartz S, Friedberg I, Ivanov IV, Davidson LA, Goldsby JS, Dahl DB, et al. A metagenomic study of diet-dependent interaction between gut microbiota and host in infants reveals differences in immune response. Genome Biol. 2012;13(4): r32.PubMedPubMedCentralCrossRef Schwartz S, Friedberg I, Ivanov IV, Davidson LA, Goldsby JS, Dahl DB, et al. A metagenomic study of diet-dependent interaction between gut microbiota and host in infants reveals differences in immune response. Genome Biol. 2012;13(4): r32.PubMedPubMedCentralCrossRef
39.
go back to reference Levy E, Rizwan Y, Thibault L, Lepage G, Brunet S, Bouthillier L, et al. Altered lipid profile, lipoprotein composition, and oxidant and antioxidant status in pediatric Crohn disease. Am J Clin Nutr. 2000;71(3):807–15.PubMedCrossRef Levy E, Rizwan Y, Thibault L, Lepage G, Brunet S, Bouthillier L, et al. Altered lipid profile, lipoprotein composition, and oxidant and antioxidant status in pediatric Crohn disease. Am J Clin Nutr. 2000;71(3):807–15.PubMedCrossRef
40.
go back to reference Haberman Y, Tickle TL, Dexheimer PJ, Kim MO, Tang D, Karns R, et al. Pediatric Crohn disease patients exhibit specific ileal transcriptome and microbiome signature. J Clin Investig. 2014;124(8):3617–33.PubMedPubMedCentralCrossRef Haberman Y, Tickle TL, Dexheimer PJ, Kim MO, Tang D, Karns R, et al. Pediatric Crohn disease patients exhibit specific ileal transcriptome and microbiome signature. J Clin Investig. 2014;124(8):3617–33.PubMedPubMedCentralCrossRef
41.
go back to reference Grasberger H, Refetoff S. Identification of the maturation factor for dual oxidase. Evolution of an eukaryotic operon equivalent. J Biol Chem. 2006;281(27):18269–72.PubMedCrossRef Grasberger H, Refetoff S. Identification of the maturation factor for dual oxidase. Evolution of an eukaryotic operon equivalent. J Biol Chem. 2006;281(27):18269–72.PubMedCrossRef
42.
go back to reference Bae YS, Choi MK, Lee WJ. Dual oxidase in mucosal immunity and host-microbe homeostasis. Trends Immunol. 2010;31(7):278–87.PubMedCrossRef Bae YS, Choi MK, Lee WJ. Dual oxidase in mucosal immunity and host-microbe homeostasis. Trends Immunol. 2010;31(7):278–87.PubMedCrossRef
43.
go back to reference Lloyd-Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila-Pacheco J, Poon TW, et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569(7758):655–62.PubMedPubMedCentralCrossRef Lloyd-Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila-Pacheco J, Poon TW, et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569(7758):655–62.PubMedPubMedCentralCrossRef
44.
go back to reference Hong SN, Joung JG, Bae JS, Lee CS, Koo JS, Park SJ, et al. RNA-seq reveals transcriptomic differences in inflamed and noninflamed intestinal mucosa of Crohn’s disease patients compared with normal mucosa of healthy controls. Inflamm Bowel Dis. 2017;23(7):1098–108.PubMedCrossRef Hong SN, Joung JG, Bae JS, Lee CS, Koo JS, Park SJ, et al. RNA-seq reveals transcriptomic differences in inflamed and noninflamed intestinal mucosa of Crohn’s disease patients compared with normal mucosa of healthy controls. Inflamm Bowel Dis. 2017;23(7):1098–108.PubMedCrossRef
45.
go back to reference Eliadou E, Moleiro J, Ribaldone DG, Astegiano M, Rothfuss K, Taxonera C, et al. Interstitial and granulomatous lung disease in inflammatory bowel disease patients. J Crohns Colitis. 2020;14(4):480–9.PubMedCrossRef Eliadou E, Moleiro J, Ribaldone DG, Astegiano M, Rothfuss K, Taxonera C, et al. Interstitial and granulomatous lung disease in inflammatory bowel disease patients. J Crohns Colitis. 2020;14(4):480–9.PubMedCrossRef
46.
go back to reference Abbasian J, Martin TM, Patel S, Tessler HH, Goldstein DA. Immunologic and genetic markers in patients with idiopathic ocular inflammation and a family history of inflammatory bowel disease. Am J Ophthalmol. 2012;154(1):72–7.PubMedPubMedCentralCrossRef Abbasian J, Martin TM, Patel S, Tessler HH, Goldstein DA. Immunologic and genetic markers in patients with idiopathic ocular inflammation and a family history of inflammatory bowel disease. Am J Ophthalmol. 2012;154(1):72–7.PubMedPubMedCentralCrossRef
47.
go back to reference Taleban S, Li D, Targan SR, Ippoliti A, Brant SR, Cho JH, et al. Ocular manifestations in inflammatory bowel disease are associated with other extra-intestinal manifestations, gender, and genes implicated in other immune-related traits. J Crohns Colitis. 2016;10(1):43–9.PubMedCrossRef Taleban S, Li D, Targan SR, Ippoliti A, Brant SR, Cho JH, et al. Ocular manifestations in inflammatory bowel disease are associated with other extra-intestinal manifestations, gender, and genes implicated in other immune-related traits. J Crohns Colitis. 2016;10(1):43–9.PubMedCrossRef
48.
go back to reference Sange AH, Srinivas N, Sarnaik MK, Modi S, Pisipati Y, Vaidya S, et al. Extra-intestinal manifestations of inflammatory bowel disease. Cureus. 2021;13(8): e17187.PubMedPubMedCentral Sange AH, Srinivas N, Sarnaik MK, Modi S, Pisipati Y, Vaidya S, et al. Extra-intestinal manifestations of inflammatory bowel disease. Cureus. 2021;13(8): e17187.PubMedPubMedCentral
49.
go back to reference Ribaldone DG, Brigo S, Mangia M, Saracco GM, Astegiano M, Pellicano R. Oral manifestations of inflammatory bowel disease and the role of non-invasive surrogate markers of disease activity. Medicines (Basel, Switzerland). 2020;7(6):33.PubMedPubMedCentral Ribaldone DG, Brigo S, Mangia M, Saracco GM, Astegiano M, Pellicano R. Oral manifestations of inflammatory bowel disease and the role of non-invasive surrogate markers of disease activity. Medicines (Basel, Switzerland). 2020;7(6):33.PubMedPubMedCentral
50.
go back to reference Rogler G, Singh A, Kavanaugh A, Rubin DT. Extraintestinal manifestations of inflammatory bowel disease: current concepts, treatment, and implications for disease management. Gastroenterology. 2021;161(4):1118–32.PubMedCrossRef Rogler G, Singh A, Kavanaugh A, Rubin DT. Extraintestinal manifestations of inflammatory bowel disease: current concepts, treatment, and implications for disease management. Gastroenterology. 2021;161(4):1118–32.PubMedCrossRef
51.
52.
go back to reference Diosdado B, van Bakel H, Strengman E, Franke L, van Oort E, Mulder CJ, et al. Neutrophil recruitment and barrier impairment in celiac disease: a genomic study. Clin Gastroenterol Hepatol. 2007;5(5):574–81.PubMedCrossRef Diosdado B, van Bakel H, Strengman E, Franke L, van Oort E, Mulder CJ, et al. Neutrophil recruitment and barrier impairment in celiac disease: a genomic study. Clin Gastroenterol Hepatol. 2007;5(5):574–81.PubMedCrossRef
53.
go back to reference Leal RF, Planell N, Kajekar R, Lozano JJ, Ordás I, Dotti I, et al. Identification of inflammatory mediators in patients with Crohn’s disease unresponsive to anti-TNFα therapy. Gut. 2015;64(2):233–42.PubMedCrossRef Leal RF, Planell N, Kajekar R, Lozano JJ, Ordás I, Dotti I, et al. Identification of inflammatory mediators in patients with Crohn’s disease unresponsive to anti-TNFα therapy. Gut. 2015;64(2):233–42.PubMedCrossRef
54.
go back to reference Tardif MR, Chapeton-Montes JA, Posvandzic A, Pagé N, Gilbert C, Tessier PA. Secretion of S100A8, S100A9, and S100A12 by neutrophils involves reactive oxygen species and potassium efflux. J Immunol Res. 2015;2015: 296149.PubMedPubMedCentralCrossRef Tardif MR, Chapeton-Montes JA, Posvandzic A, Pagé N, Gilbert C, Tessier PA. Secretion of S100A8, S100A9, and S100A12 by neutrophils involves reactive oxygen species and potassium efflux. J Immunol Res. 2015;2015: 296149.PubMedPubMedCentralCrossRef
55.
go back to reference Simard JC, Cesaro A, Chapeton-Montes J, Tardif M, Antoine F, Girard D, et al. S100A8 and S100A9 induce cytokine expression and regulate the NLRP3 inflammasome via ROS-dependent activation of NF-κB(1.). PLoS ONE. 2013;8(8):e72138.PubMedPubMedCentralCrossRef Simard JC, Cesaro A, Chapeton-Montes J, Tardif M, Antoine F, Girard D, et al. S100A8 and S100A9 induce cytokine expression and regulate the NLRP3 inflammasome via ROS-dependent activation of NF-κB(1.). PLoS ONE. 2013;8(8):e72138.PubMedPubMedCentralCrossRef
56.
go back to reference Manolakis AC, Kapsoritakis AN, Tiaka EK, Potamianos SP. Calprotectin, calgranulin C, and other members of the s100 protein family in inflammatory bowel disease. Dig Dis Sci. 2011;56(6):1601–11.PubMedCrossRef Manolakis AC, Kapsoritakis AN, Tiaka EK, Potamianos SP. Calprotectin, calgranulin C, and other members of the s100 protein family in inflammatory bowel disease. Dig Dis Sci. 2011;56(6):1601–11.PubMedCrossRef
57.
go back to reference Stallhofer J, Friedrich M, Konrad-Zerna A, Wetzke M, Lohse P, Glas J, et al. Lipocalin-2 Is a disease activity marker in inflammatory bowel disease regulated by IL-17A, IL-22, and TNF-α and modulated by IL23R genotype status. Inflamm Bowel Dis. 2015;21(10):2327–40.PubMed Stallhofer J, Friedrich M, Konrad-Zerna A, Wetzke M, Lohse P, Glas J, et al. Lipocalin-2 Is a disease activity marker in inflammatory bowel disease regulated by IL-17A, IL-22, and TNF-α and modulated by IL23R genotype status. Inflamm Bowel Dis. 2015;21(10):2327–40.PubMed
58.
go back to reference Abella V, Scotece M, Conde J, Gómez R, Lois A, Pino J, et al. The potential of lipocalin-2/NGAL as biomarker for inflammatory and metabolic diseases. Biomarkers. 2015;20(8):565–71.PubMedPubMedCentralCrossRef Abella V, Scotece M, Conde J, Gómez R, Lois A, Pino J, et al. The potential of lipocalin-2/NGAL as biomarker for inflammatory and metabolic diseases. Biomarkers. 2015;20(8):565–71.PubMedPubMedCentralCrossRef
59.
go back to reference Xiao X, Yeoh BS, Vijay-Kumar M. Lipocalin 2: an emerging player in iron homeostasis and inflammation. Annu Rev Nutr. 2017;37:103–30.PubMedCrossRef Xiao X, Yeoh BS, Vijay-Kumar M. Lipocalin 2: an emerging player in iron homeostasis and inflammation. Annu Rev Nutr. 2017;37:103–30.PubMedCrossRef
60.
go back to reference Playford RJ, Belo A, Poulsom R, Fitzgerald AJ, Harris K, Pawluczyk I, et al. Effects of mouse and human lipocalin homologues 24p3/lcn2 and neutrophil gelatinase-associated lipocalin on gastrointestinal mucosal integrity and repair. Gastroenterology. 2006;131(3):809–17.PubMedCrossRef Playford RJ, Belo A, Poulsom R, Fitzgerald AJ, Harris K, Pawluczyk I, et al. Effects of mouse and human lipocalin homologues 24p3/lcn2 and neutrophil gelatinase-associated lipocalin on gastrointestinal mucosal integrity and repair. Gastroenterology. 2006;131(3):809–17.PubMedCrossRef
61.
go back to reference Chu H, Pazgier M, Jung G, Nuccio SP, Castillo PA, de Jong MF, et al. Human α-defensin 6 promotes mucosal innate immunity through self-assembled peptide nanonets. Science (New York, NY). 2012;337(6093):477–81.CrossRef Chu H, Pazgier M, Jung G, Nuccio SP, Castillo PA, de Jong MF, et al. Human α-defensin 6 promotes mucosal innate immunity through self-assembled peptide nanonets. Science (New York, NY). 2012;337(6093):477–81.CrossRef
63.
go back to reference Hayashi R, Tsuchiya K, Fukushima K, Horita N, Hibiya S, Kitagaki K, et al. Reduced human α-defensin 6 in noninflamed Jejunal tissue of patients with Crohn’s disease. Inflamm Bowel Dis. 2016;22(5):1119–28.PubMedCrossRef Hayashi R, Tsuchiya K, Fukushima K, Horita N, Hibiya S, Kitagaki K, et al. Reduced human α-defensin 6 in noninflamed Jejunal tissue of patients with Crohn’s disease. Inflamm Bowel Dis. 2016;22(5):1119–28.PubMedCrossRef
64.
go back to reference Ye Y, Xiao L, Wang SJ, Yue W, Yin QS, Sun MY, et al. Up-regulation of REG3A in colorectal cancer cells confers proliferation and correlates with colorectal cancer risk. Oncotarget. 2016;7(4):3921–33.PubMedCrossRef Ye Y, Xiao L, Wang SJ, Yue W, Yin QS, Sun MY, et al. Up-regulation of REG3A in colorectal cancer cells confers proliferation and correlates with colorectal cancer risk. Oncotarget. 2016;7(4):3921–33.PubMedCrossRef
65.
go back to reference Marafini I, Di Sabatino A, Zorzi F, Monteleone I, Sedda S, Cupi ML, et al. Serum regenerating islet-derived 3-alpha is a biomarker of mucosal enteropathies. Aliment Pharmacol Ther. 2014;40(8):974–81.PubMedCrossRef Marafini I, Di Sabatino A, Zorzi F, Monteleone I, Sedda S, Cupi ML, et al. Serum regenerating islet-derived 3-alpha is a biomarker of mucosal enteropathies. Aliment Pharmacol Ther. 2014;40(8):974–81.PubMedCrossRef
66.
go back to reference Nunes T, Etchevers MJ, Sandi MJ, Pinó Donnay S, Grandjean T, Pellisé M, et al. Pancreatitis-associated protein does not predict disease relapse in inflammatory bowel disease patients. PLoS ONE. 2014;9(1): e84957.PubMedPubMedCentralCrossRef Nunes T, Etchevers MJ, Sandi MJ, Pinó Donnay S, Grandjean T, Pellisé M, et al. Pancreatitis-associated protein does not predict disease relapse in inflammatory bowel disease patients. PLoS ONE. 2014;9(1): e84957.PubMedPubMedCentralCrossRef
67.
go back to reference Luo S, Surapaneni A, Zheng Z, Rhee EP, Coresh J, Hung AM, et al. NAT8 variants, N-acetylated amino acids, and progression of CKD. CJASN. 2020;16(1):37–47.PubMedPubMedCentralCrossRef Luo S, Surapaneni A, Zheng Z, Rhee EP, Coresh J, Hung AM, et al. NAT8 variants, N-acetylated amino acids, and progression of CKD. CJASN. 2020;16(1):37–47.PubMedPubMedCentralCrossRef
68.
go back to reference Jiang H, Tang E, Chen Y, Liu H, Zhao Y, Lin M, et al. Squalene synthase predicts poor prognosis in stage I–III colon adenocarcinoma and synergizes squalene epoxidase to promote tumor progression. Cancer Sci. 2022;113(3):971–85.PubMedPubMedCentralCrossRef Jiang H, Tang E, Chen Y, Liu H, Zhao Y, Lin M, et al. Squalene synthase predicts poor prognosis in stage I–III colon adenocarcinoma and synergizes squalene epoxidase to promote tumor progression. Cancer Sci. 2022;113(3):971–85.PubMedPubMedCentralCrossRef
Metadata
Title
Predicting diagnostic biomarkers associated with immune infiltration in Crohn's disease based on machine learning and bioinformatics
Authors
Wenhui Bao
Lin Wang
Xiaoxiao Liu
Ming Li
Publication date
01-12-2023
Publisher
BioMed Central
Published in
European Journal of Medical Research / Issue 1/2023
Electronic ISSN: 2047-783X
DOI
https://doi.org/10.1186/s40001-023-01200-9

Other articles of this Issue 1/2023

European Journal of Medical Research 1/2023 Go to the issue